EP2519538A1 - Cyclosporin-analoga - Google Patents

Cyclosporin-analoga

Info

Publication number
EP2519538A1
EP2519538A1 EP10799265A EP10799265A EP2519538A1 EP 2519538 A1 EP2519538 A1 EP 2519538A1 EP 10799265 A EP10799265 A EP 10799265A EP 10799265 A EP10799265 A EP 10799265A EP 2519538 A1 EP2519538 A1 EP 2519538A1
Authority
EP
European Patent Office
Prior art keywords
carbon atoms
compound
optionally substituted
branched
same
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP10799265A
Other languages
English (en)
French (fr)
Inventor
Cyprian Okwara Ogbu
Thomas Edward Richardson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Scynexis Chemistry and Automation Inc
Original Assignee
Scynexis Chemistry and Automation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Scynexis Chemistry and Automation Inc filed Critical Scynexis Chemistry and Automation Inc
Publication of EP2519538A1 publication Critical patent/EP2519538A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • C07K7/645Cyclosporins; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to novel compounds, compositions containing them, processes for their preparation, and their use as therapeutics, for example as antiviral agents.
  • Cyclosporine A is well known for its immunosuppressive activity and a range of therapeutic uses, including antifungal, anti-parasitic, and anti-inflammatory as well as anti-HIV activity. Cyclosporine A and certain derivatives have been reported as having anti-HCV activity, see Watashi et al., Hepatology, 2003, Vol. 38, pp 1282-1288,
  • Cyclosporine A (cyclosporine) derivatives modified in the 4-position to introduce hydroxyl are known in the literature.
  • [4'-Hydroxy-N- methylleucine] 4 cyclosporine A is disclosed in European Patent No. 484,281, and is stated to be active against HIV-1 replication.
  • 3-Ether/thioether-[4'-hydroxy- N-methylleucine] 4 cyclosporine A derivatives are described in U.S. Patent Nos.
  • the invention provides a cyclosporine derivative of the formula (I):
  • B represents ethyl, 1-hydroxyethyl, isopropyl or n-propyl
  • R 1 represents:
  • straight- or branched- chain alkyl having from one to six carbon atoms; straight- or branched- chain alkenyl having from two to six carbon atoms;
  • R 2 represents:- straight- or branched- chain alkyl having from one to six carbon atoms, optionally substituted by one or more groups R 4 which are the same or different;
  • cycloalkyl having from three to six ring carbon atoms optionally substituted by one or more substituents which are the same or different selected from the group consisting of halogen, hydroxy, amino, N-monoalkylamino and
  • R 3 represents:
  • N-monoalkylamino and N,N-dialkylamino straight- or branched- chain alkynyl having from two to six carbon atoms, optionally substituted by one or more substituents which are the same or different selected from the group consisting of halogen, hydroxy, amino,
  • cycloalkyl having from three to six ring carbon atoms optionally substituted by one or more substituents which are the same or different selected from the group consisting of halogen, hydroxy, amino, N-monoalkylamino and
  • R 4 is selected from the group consisting of halogen; hydroxy; alkoxy;
  • R 5 and R 6 which are the same or different, each represent:
  • cycloalkyl having from three to six ring carbon atoms optionally substituted by straight- or branched- chain alkyl having from one to six carbon atoms; phenyl optionally substituted by from one to five substituents which are the same or different selected from the group consisting of halogen, alkoxy, cyano, alkoxycarbonyl, amino, alkylamino and dialkylamino;
  • heterocyclic ring which is saturated or unsaturated having five or six ring atoms of which from one to three are heteroatoms which are the same or different selected from nitrogen, sulfur and oxygen, which heterocyclic ring is optionally substituted by one or more substituents which are the same or different selected from the group consisting of halogen, alkoxy, cyano, alkoxycarbonyl, amino, alkylamino and dialkylamino;
  • R 5 and R 6 together with the nitrogen atom to which they are attached, form a saturated or unsaturated heterocyclic ring having from four to six ring atoms, which ring optionally has another ring heteroatom selected from the group consisting of nitrogen, oxygen and sulfur and is optionally substituted by from one to four substituents which are the same or different selected from the group consisting of alkyl, phenyl and benzyl;
  • R 7 represents hydrogen; straight- or branched- chain alkyl having from one to six carbon atoms; or phenyl optionally substituted by from one to five substituents which are the same or different selected from the group consisting of halogen and alkoxy;
  • p is zero, one or two;
  • m is an integer from two to four;
  • substituents A, B, R 1 and R 2 may contribute to optical and/or stereoisomerism. All such forms are embraced by the present invention.
  • compositions comprising a compound provided herein along with a pharmaceutically acceptable excipient, carrier or diluent.
  • compositions provided herein.
  • pharmaceutically acceptable salts include salts with alkali metals, e.g., sodium, potassium or lithium, or with alkaline-earth metals, e.g., magnesium or calcium, the ammonium salt or the salts of nitrogenous bases, e.g., ethanolamine, diethanolamine, trimethylamine, triethylamine, methylamine,
  • dicyclohexylamine N-benzylphenethylamine, N,N'-dibenzylethylenediamine, diphenylenediamine, benzhydrylamine, quinine, choline, arginine, lysine, leucine or dibenzylamine.
  • kits for using a compound or composition provided herein to treat or prevent an infection, a neurodegenerative disease, ischemia/reperfusion damage, an inflammatory disease or an autoimmune disease generally comprise administering to a subject having the condition or disease an amount of the compound or composition effective to treat or prevent the disease or condition.
  • Exemplary infections include HCV or HIV infection and others described in detail herein.
  • a compound or composition described herein for use in therapy In another aspect, provided is a compound or composition described herein for use in treatment or prevention of an infection, a neurodegenerative disease,
  • ischemia/reperfusion damage an inflammatory disease or an autoimmune disease.
  • uses of compounds or compositions in the manufacture of a medicament in another aspect, provided is a compound or composition described herein for use in the manufacture of a medicament for treatment or prevention of an infection, a neurodegenerative disease, ischemia/reperfusion damage, an inflammatory disease or an autoimmune disease.
  • Cyclosporine refers to any cyclosporine compound known to those of skill in the art, or a derivative thereof. See, e.g., Ruegger et al, 1976, Helv. Chim. Acta. 59: 1075-92; Borel et ah, 1977, Immunology 32: 1017-25; the contents of which are hereby incorporated by reference in their entireties.
  • Exemplary compounds of the invention are cyclosporine derivatives. Unless noted otherwise, a cyclosporine described herein is a cyclosporine A, and a cyclosporine derivative described herein is a derivative of cyclosporine A.
  • Alkyl refers to monovalent saturated aliphatic hydrocarbyl groups particularly having up to 1 1 carbon atoms, more particularly as a lower alkyl, from 1 to 8 carbon atoms and still more particularly, from 1 to 6 carbon atoms.
  • the hydrocarbon chain may be either straight-chained or branched. This term is exemplified by groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, tert-butyl, n-hexyl, n-octyl, tert-octyl and the like.
  • Alkylene refers to divalent saturated aliphatic hydrocarbyl groups particularly having up to 1 1 carbon atoms and more particularly 1 to 6 carbon atoms which can be straight-chained or branched. This term is exemplified by groups such as methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), the propylene isomers (e.g., -CH 2 CH 2 CH 2 - and -CH(CH 3 )CH 2 -) and the like.
  • alkenyl refers to monovalent olefinically unsaturated hydrocarbyl groups, in one embodiment, having up to 1 1 carbon atoms, in another embodiment, from 2 to 8 carbon atoms, and in yet another embodiment, from 2 to 6 carbon atoms, which can be straight-chained or branched and having at least 1 or from 1 to 2 sites of olefinic unsaturation.
  • Alkynyl refers to acetylenically unsaturated hydrocarbyl groups particularly having up to 1 1 carbon atoms and more particularly 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 and particularly from 1 to 2 sites of alkynyl unsaturation.
  • alkynyl groups include acetylenic, ethynyl (-C ⁇ CH), propargyl (-CH 2 C ⁇ CH), and the like.
  • Alkoxy refers to the group -OR where R is alkyl.
  • the alkyl group has up to 1 1 carbon atoms, more particularly as a lower alkyl, from 1 to 8 carbon atoms and still more particularly, from 1 to 6 carbon atoms.
  • Particular alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n- pentoxy, n-hexoxy, 1 ,2-dimethylbutoxy, and the like.
  • N-Monoalkylamino refers to the group H-NR'-, wherein R' is selected from hydrogen and alkyl.
  • the alkyl group has up to 1 1 carbon atoms, more particularly as a lower alkyl, from 1 to 8 carbon atoms and still more particularly, from 1 to 6 carbon atoms.
  • Aryl refers to an optionally substituted aromatic hydrocarbon radical, for example phenyl.
  • Arylamino refers to the group aryl-NR'-, wherein R' is selected from hydrogen, aryl and heteroaryl.
  • Bmt refers to 2(S)-amino-3(R)-hydroxy-4(R)- methyl-6(E)-octenoic acid.
  • ⁇ , ⁇ -Dialkylamino means a radical -NRR' where R and R' independently represent an alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroaryl, or substituted heteroaryl group as defined herein.
  • Halogen or "halo” refers to chloro, bromo, fluoro or iodo.
  • Heteroaryl refers to an optionally substituted saturated or unsaturated heterocyclic radical, of which from 1 to 3 are hetero ring atoms selected from the group consisting of sulfur, oxygen and nitrogen. Generally the heterocyclic ring has from 4 to 7 ring atoms, e.g. 5 or 6 ring atoms.
  • heteroaryl examples include thienyl, furyl, pyrrolyl, oxazinyl, thiazinyl, pyrazinyl, pyrimidinyl, pyridazinyl, thiazolyl, oxazolyl, imidazolyl, morpholinyl, pyrazolyl, tetrahydorfuryl oxadiazolyl, thiadiazolyl and isoxazolyl.
  • Haldroxy refers to the radical -OH.
  • Thioalkyl refers to the group -SR where R is alkyl.
  • the alkyl group has up to 1 1 carbon atoms, more particularly as a lower alkyl, from 1 to 8 carbon atoms and still more particularly, from 1 to 6 carbon atoms. Examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, and the like.
  • “Pharmaceutically acceptable salt” refers to any salt of a compound of this invention which retains its biological properties and which is not toxic or otherwise undesirable for pharmaceutical use. Such salts may be derived from a variety of organic and inorganic counter-ions well known in the art.
  • Such salts include: (1) acid addition salts formed with organic or inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, sulfamic, acetic, trifluoroacetic, trichloroacetic, propionic, hexanoic, cyclopentylpropionic, glycolic, glutaric, pyruvic, lactic, malonic, succinic, sorbic, ascorbic, malic, maleic, fumaric, tartaric, citric, benzoic, 3-(4-hydroxybenzoyl)benzoic, picric, cinnamic, mandelic, phthalic, lauric, methanesulfonic, ethanesulfonic, 1,2-ethane- disulfonic, 2-hydroxyethanesulfonic, benzenesulfonic, 4-chlorobenzenesulfonic, 2- naphthalenesulfonic, 4-toluenesul
  • Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium and the like, and when the compound contains a basic functionality, salts of non-toxic organic or inorganic acids, such as hydrohalides, e.g.
  • physiologically acceptable cation refers to a non-toxic, physiologically acceptable cationic counterion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium and tetraalkylammonium cations and the like.
  • Solvate refers to a compound of the present invention or a salt thereof that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non- covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
  • enantiomers and those that are non-superimposable mirror images of each other are termed “enantiomers".
  • An enantiomer can be characterized by the absolute configuration of its asymmetric center and is designated (R) or (S) according to the rules of Cahn and Prelog (Cahn et al, 1966, Angew. Chem. 78:413-447, Angew. Chem., Int. Ed. Engl. 5:385-414 (errata: Angew. Chem., Int. Ed. Engl.
  • a chiral compound can exist as either individual enantiomer or as a mixture thereof.
  • a mixture containing equal proportions of enantiomers is called a "racemic mixture”.
  • the compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as the individual (R)- or (5)-enantiomer or as a mixture thereof.
  • the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof.
  • Methods for determination of stereochemistry and separation of stereoisomers are well-known in the art.
  • the present invention provides the stereoisomers of the compounds depicted herein upon treatment with base.
  • the compounds of the invention are N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • stereochemically pure A stereochemically pure compound has a level of
  • stereochemically pure designates a compound that is substantially free of alternate isomers.
  • the compound is 85%, 90%>, 91 >, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or 99.9% free of other isomers.
  • the terms “subject” and “patient” are used interchangeably herein.
  • the terms “subject” and “subjects” refer to an animal, in some embodiments, a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey such as a cynomolgous monkey, a chimpanzee and a human), and a human.
  • the subject is a farm animal (e.g., a horse, a cow, a pig, etc.) or a pet (e.g., a dog or a cat).
  • the subject is a human.
  • a therapeutic agent refers to any agent(s) which can be used in the treatment, management, or amelioration of a disorder or one or more symptoms thereof.
  • the term “therapeutic agent” refers to a compound of the invention.
  • the term “therapeutic agent” does not refer to a compound of the invention.
  • a therapeutic agent is an agent that is known to be useful for, or has been or is currently being used for the treatment, management, prevention, or amelioration of a disorder or one or more symptoms thereof.
  • “Therapeutically effective amount” means an amount of a compound or complex or composition that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease.
  • a “therapeutically effective amount” can vary depending on, inter alia, the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.
  • Treating” or “treatment” of any disease or disorder refers, in one
  • treating refers to ameliorating a disease or disorder that exists in a subject.
  • “treating” or “treatment” refers to ameliorating at least one physical parameter, which may be indiscernible by the subject.
  • “treating” or “treatment” refers to modulating the disease or disorder, either physically ⁇ e.g., stabilization of a discernible symptom) or physiologically ⁇ e.g., stabilization of a physical parameter) or both.
  • “treating” or “treatment” refers to delaying the onset of the disease or disorder.
  • prophylactic agent and “prophylactic agents” as used refer to any agent(s) which can be used in the prevention of a disorder or one or more symptoms thereof.
  • the term “prophylactic agent” refers to a compound of the invention.
  • the term “prophylactic agent” does not refer a compound of the invention.
  • a prophylactic agent is an agent which is known to be useful for, or has been or is currently being used to prevent or impede the onset, development, progression and/or severity of a disorder.
  • the terms “prevent”, “preventing” and “prevention” refer to the prevention of the recurrence, onset, or development of one or more symptoms of a disorder in a subject resulting from the administration of a therapy ⁇ e.g., a prophylactic or therapeutic agent), or the administration of a combination of therapies (e.g., a
  • prophylactically effective amount refers to the amount of a therapy (e.g., prophylactic agent) which is sufficient to result in the prevention of the development, recurrence or onset of one or more symptoms associated with a disorder, or to enhance or improve the prophylactic effect(s) of another therapy (e.g., another prophylactic agent).
  • a therapy e.g., prophylactic agent
  • another therapy e.g., another prophylactic agent
  • label refers to a display of written, printed or graphic matter upon the immediate container of an article, for example the written material displayed on a vial containing a pharmaceutically active agent.
  • labeling refers to all labels and other written, printed or graphic matter upon any article or any of its containers or wrappers or accompanying such article, for example, a package insert or instructional videotapes or DVDs accompanying or associated with a container of a pharmaceutically active agent.
  • B represents ethyl
  • R 1 represents hydrogen. In another embodiment R 1 represents straight- or branched- chain alkyl having from one to six carbon atoms;
  • R 1 represents -XR 3 .
  • R 1 represents methyl.
  • R 3 represents straight- or branched- chain alkyl having from one to six carbon atoms. In a further embodiment R 3 represents methyl or ethyl. In a further embodiment R 3 represents straight- or branched- chain alkyl having from one to six carbon atoms substituted by a group R 4 . In a further embodiment R 3 represents straight- or branched- chain alkyl having from one to four carbon atoms substituted by a group R 4 . In a further embodiment R 3 represents straight- or branched- chain alkyl having from one to four carbon atoms substituted by ⁇ , ⁇ -dialkylamino. In a further embodiment R 3 represents ethyl substituted by N,N-dimethylamino.
  • R 2 represents straight- or branched- chain alkyl having from one to six carbon atoms, optionally substituted by one or more groups R 4 which are the same or different.
  • R 2 represents straight- or branched- chain alkyl having from one to four carbon atoms, optionally substituted by a group R 4 .
  • R 2 represents straight- or branched- chain alkyl having one or two carbon atoms, optionally substituted by a group R 4 .
  • R 2 represents, methyl, ethyl, or ethyl substituted by a group R 4 .
  • R 4 represents phenyl optionally substituted by one or more substituents which are the same or different selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, cyano, amino, N-alkylamino and ⁇ , ⁇ -dialkylamino. In a further embodiment R 4 represents phenyl.
  • X is oxygen or sulfur. In certain embodiments, X is oxygen or sulfur. In further embodiments X is oxygen. In still further embodiments, X is sulfur.
  • the compound provided herein is selected from the following:
  • compounds of formula (I) can be prepared by the reaction of a compound of formula (II):
  • the reaction generally takes place a temperature of from about 0°C to about 100°C, such as from about 0°C to about 50°C (for example at room temperature) in a solvent, which may be the compound of formula R 2 OH.
  • the reaction may be performed in the presence of an acid, for example a Bronsted acid or a Lewis acid, or an acid such as p-toluenesulfonic acid or methanesulfonic acid.
  • the compound of formula CH(OR 2 ) 3 may be a trialkylorthoformate, for example triethylortho formate.
  • compounds of formula (I) can be prepared by the reaction of a compound of formula (II) as defined above with a salt comprising a cation of formula (III):
  • Examples of suitable salts for this reaction include salts of strong acids such as a triflate, trifluoracetate or trichloroacetate salt.
  • the reaction is typically performed in the presence of a buffering agent such as magnesium oxide in a solvent such as an aromatic hydrocarbon, e.g. toluene or ⁇ , ⁇ , ⁇ -trifluorotoluene.
  • a buffering agent such as magnesium oxide
  • a solvent such as an aromatic hydrocarbon, e.g. toluene or ⁇ , ⁇ , ⁇ -trifluorotoluene.
  • the reaction is generally performed at a temperature from about 0°C to about 100°C, such as from about 50°C to about 100°C, e.g. from about 80°C to about 85°C.
  • compounds of formula (I) in which R 1 represents XR 3 can be prepared by treating the corresponding compound of formula (I) in which R 1 represents hydrogen with a base in an appropriate solvent to generate a polyanionic species, followed by the reaction of the polyanion thus obtained with a electrophile of formula R 3 X-L, wherein R 3 and X are as defined above and L is a leaving group.
  • the compound of formula (I) in which R 1 is hydrogen is dissolved in an appropriate solvent and cooled to about -70°C.
  • Solvents include tetrahydrofuran, dimethyoxymethane, methyl tert-butylether, dioxane, and the like.
  • the resulting mixture is generally allowed to react for about 1 hour and is optionally allowed to warm to about -20°C.
  • the reaction mixture is typically cooled to about -70°C and an appropriate electrophile is added.
  • Preferred bases for this reaction include n-butyl lithium, lithium diisopropylamide, lithium diisopropylamide in combination with lithium chloride and sodium amide.
  • Suitable electrophiles include, but are not limited to activated alkyl and alkenyl halides or sulfonates, disulfides,
  • Compounds of formula (II) can be prepared according to methods known to one of skill in the art, for example, methods described in U.S. Patent Nos. 5,948,884; 5,994,299 and 6,583,265 and in PCT publication nos. W099/32512 and WO99/67280. The contents of these references are hereby incorporated by reference in their entireties.
  • Compounds of formula (III) are known or can be prepared by the application and adaptation of known methods.
  • a cyclosporine compound of the invention can be in a neutral form, or in a salt form.
  • the salt form can be any salt form known to those of skill in the art. Particularly useful salt forms are those that are coordinated with phosphate, citrate, acetate, chloride, methanesulfonate or propionate.
  • an acid addition salt can be formed.
  • the acid which can be used to prepare an acid addition salt includes that which produces, when combined with the free base, a pharmaceutically acceptable salt, that is, a salt whose anion is non-toxic to a subject in the pharmaceutical doses of the salt.
  • salts within the scope of the invention are those derived from the following acids: mineral acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, sulfamic acid and nitric acid; and organic acids such as acetic, trifluoroacetic, trichloroacetic, propionic, hexanoic, cyclopentylpropionic, glycolic, glutaric, pyruvic, lactic, malonic, succinic, sorbic, ascorbic, malic, maleic, fumaric, tartaric, citric, benzoic, 3-(4- hydroxybenzoyl)benzoic, picric, cinnamic, mandelic, phthalic, lauric, methanesulfonic, ethanesulfonic, 1 ,2-ethane-disulfonic, 2-hydroxyethanesulfonic, benzenesulfonic, 4- chlorobenzenesulfonic, 2-na
  • camphorsulfonic 4-methylbicyclo[2.2.2]-oct-2-ene-l-carboxylic, glucoheptonic, 3- phenylpropionic, trimethylacetic, tert-butylacetic, lauryl sulfuric, gluconic, benzoic, glutamic, hydroxynaphthoic, salicylic, stearic, cyclohexylsulfamic, quinic, muconic acid and the like acids.
  • the corresponding acid addition salts include hydrohalides, e.g. hydrochloride and hydrobromide, sulfate, phosphate, sulfamate, nitrate, acetate, trifluoroacetate, trichloroacetate, propionate, hexanoate, cyclopentylpropionate, glycolate, glutarate, pyruvate, lactate, malonate, succinate, sorbate, ascorbate, malate, maleate, fumarate, tartarate, citrate, benzoate, 3-(4-hydroxybenzoyl)benzoate, picrate, cinnamate, mandelate, phthalate, laurate, methanesulfonate (mesylate), ethanesulfonate, 1 ,2-ethanedisulfonate, 2-hydroxyethanesulfonate, benzenesulfonate (besylate), 4-chlorobenzenesulfon
  • acid addition salts of the compounds of this invention can be prepared by reaction of the free base with the appropriate acid, by the application or adaptation of known methods.
  • the acid addition salts of the compounds of this invention can be prepared either by dissolving the free base in aqueous or aqueous-alcohol solution or other suitable solvents containing the appropriate acid and isolating the salt by evaporating the solution, or by reacting the free base and acid in an organic solvent, in which case the salt separates directly or can be obtained by concentration of the solution.
  • the acid addition salts of the compounds of this invention can be regenerated from the salts by the application or adaptation of known methods.
  • parent compounds of the invention can be regenerated from their acid addition salts by treatment with an alkali, e.g., aqueous sodium bicarbonate solution or aqueous ammonia solution.
  • an alkali e.g., aqueous sodium bicarbonate solution or aqueous ammonia solution.
  • base addition salts can be formed.
  • salts including for example alkali and alkaline earth metal salts, within the scope of the invention are those derived from the following bases: sodium hydride, sodium hydroxide, potassium hydroxide, calcium hydroxide, magnesium hydroxide, aluminum hydroxide, lithium hydroxide, zinc hydroxide, barium hydroxide, and organic amines such as aliphatic, alicyclic, or aromatic organic amines, such as ammonia, methylamine, dimethylamine, diethylamine, picoline, ethanolamine, diethanolamine, triethanolamine, ethylenediamine, lysine, arginine, ornithine, choline, ⁇ , ⁇ '-dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, N-methylglucamine piperazine, tris(hydroxymethyl)- aminomethane, tetramethylammonium hydroxide, and the like.
  • bases sodium hydride, sodium hydro
  • Metal salts of compounds of the present invention can be obtained by contacting a hydride, hydroxide, carbonate or similar reactive compound of the chosen metal in an aqueous or organic solvent with the free acid form of the compound.
  • the aqueous solvent employed may be water or it may be a mixture of water with an organic solvent, in certain embodiments, an alcohol such as methanol or ethanol, a ketone such as acetone, an aliphatic ether such as tetrahydrofuran, or an ester such as ethyl acetate.
  • Such reactions are normally conducted at ambient temperature but they may, if desired, be conducted with heating.
  • Amine salts of compounds of the present invention can be obtained by contacting an amine in an aqueous or organic solvent with the free acid form of the compound.
  • Suitable aqueous solvents include water and mixtures of water with alcohols such as methanol or ethanol, ethers such as tetrahydrofuran, nitriles, such as acetonitrile, or ketones such as acetone. Amino acid salts may be similarly prepared.
  • the base addition salts of the compounds of this invention can be regenerated from the salts by the application or adaptation of known methods.
  • parent compounds of the invention can be regenerated from their base addition salts by treatment with an acid, e.g., hydrochloric acid.
  • cyclosporine compounds used in the method of the present invention can be administered in certain embodiments using pharmaceutical compositions containing at least one compound of general formula (I), if appropriate in the salt form, either used alone or in the form of a combination with one or more compatible and pharmaceutically acceptable carriers, such as diluents or adjuvants, or with another anti-HCV agent.
  • pharmaceutical compositions containing at least one compound of general formula (I) if appropriate in the salt form, either used alone or in the form of a combination with one or more compatible and pharmaceutically acceptable carriers, such as diluents or adjuvants, or with another anti-HCV agent.
  • compatible and pharmaceutically acceptable carriers such as diluents or adjuvants
  • another anti-HCV agent such as diluents or adjuvants
  • cyclosporine compounds of the present invention are administered orally.
  • compositions for oral administration may be made, as solid compositions for oral administration, of tablets, pills, hard gelatin capsules, powders or granules.
  • the active product according to the invention is mixed with one or more inert diluents or adjuvants, such as sucrose, lactose or starch.
  • compositions can comprise substances other than diluents, for example a lubricant, such as magnesium stearate, or a coating intended for controlled release
  • compositions for oral administration of solutions which are pharmaceutically acceptable, suspensions, emulsions, syrups and elixirs containing inert diluents, such as water or liquid paraffin.
  • solutions which are pharmaceutically acceptable, suspensions, emulsions, syrups and elixirs containing inert diluents, such as water or liquid paraffin.
  • inert diluents such as water or liquid paraffin.
  • These compositions can also comprise substances other than diluents, for example wetting, sweetening or flavoring products.
  • compositions for parenteral administration can be emulsions or sterile solutions.
  • Use can be made, as solvent or vehicle, of propylene glycol, a polyethylene glycol, vegetable oils, in particular olive oil, or injectable organic esters, for example ethyl oleate.
  • These compositions can also contain adjuvants, in particular wetting, isotonizing, emulsifying, dispersing and stabilizing agents.
  • Sterilization can be carried out in several ways, for example using a bacteriological filter, by radiation or by heating. They can also be prepared in the form of sterile solid compositions which can be dissolved at the time of use in sterile water or any other injectable sterile medium.
  • compositions for rectal administration are suppositories or rectal capsules which contain, in addition to the active principle, excipients such as cocoa butter, semisynthetic glycerides or polyethylene glycols.
  • compositions can also be aerosols.
  • the compositions can be stable sterile solutions or solid compositions dissolved at the time of use in apyrogenic sterile water, in saline or any other pharmaceutically acceptable vehicle.
  • the active principle is finely divided and combined with a water-soluble solid diluent or vehicle, for example dextran, mannitol or lactose.
  • a composition of the invention is a pharmaceutical composition or a single unit dosage form.
  • Pharmaceutical compositions and single unit dosage forms of the invention comprise a prophylactically or therapeutically effective amount of one or more prophylactic or therapeutic agents (e.g., a compound of the invention, or other prophylactic or therapeutic agent), and typically one or more pharmaceutically acceptable carriers or excipients.
  • prophylactic or therapeutic agents e.g., a compound of the invention, or other prophylactic or therapeutic agent
  • pharmaceutically acceptable carriers or excipients typically one or more pharmaceutically acceptable carriers or excipients.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • water is a carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin.
  • Typical pharmaceutical compositions and dosage forms comprise one or more excipients.
  • Suitable excipients are well-known to those skilled in the art of pharmacy, and non limiting examples of suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a subject and the specific active ingredients in the dosage form.
  • the composition or single unit dosage form if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • Lactose free compositions of the invention can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmocopia (USP) SP (XXI)/NF (XVI).
  • USP U.S. Pharmocopia
  • XXI U.S. Pharmocopia
  • NF NF
  • lactose free compositions comprise an active ingredient, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts.
  • Exemplary lactose free dosage forms comprise an active ingredient, micro crystalline cellulose, pre-gelatinized starch, and magnesium stearate.
  • This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water is widely accepted in the pharmaceutical arts as a means of simulating long term storage in order to determine characteristics such as shelf life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80.
  • water and heat accelerate the decomposition of some compounds.
  • the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine are, in certain embodiments, anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose.
  • Such compounds which are referred to herein as
  • antioxidants include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • compositions and single unit dosage forms can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained- release formulations and the like.
  • Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc.
  • Such compositions and dosage forms will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent, in one embodiment, in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject.
  • the formulation should suit the mode of administration.
  • compositions or single unit dosage forms are sterile and in suitable form for administration to a subject, such as an animal subject, in one embodiment, a mammalian subject, such as a human subject.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, intramuscular, subcutaneous, oral, buccal, sublingual, inhalation, intranasal, transdermal, topical, transmucosal, intra-tumoral, intra-synovial and rectal administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal or topical administration to human beings.
  • a pharmaceutical composition is formulated in accordance with routine procedures for subcutaneous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocamne to ease pain at the site of the injection.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a subject, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in- water emulsions, or water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral
  • sterile solids e.g., crystalline or amorphous solids
  • liquid dosage forms suitable for parenteral administration to a subject.
  • composition, shape, and type of dosage forms of the invention will typically vary depending on their use.
  • a dosage form used in the initial treatment of viral infection may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the maintenance treatment of the same infection.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease or disorder.
  • compositions of the invention are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water- free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • Typical dosage forms of the invention comprising a compound of the invention, or a pharmaceutically acceptable salt, solvate or hydrate thereof lie within the range of from about 0.1 mg to about 1000 mg per day, given as a single once-a-day dose in the morning or in one aspect, as divided doses throughout the day taken with food.
  • dosage forms of the invention have about 0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 2.0, 2.5, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 100, 200, 250, 500 or 1000 mg of the active cyclosporine.
  • compositions of the invention that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets ⁇ e.g., chewable tablets), caplets, capsules, and liquids ⁇ e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally,
  • the oral dosage forms are solid and prepared under anhydrous conditions with anhydrous ingredients, as described in detail in the sections above.
  • anhydrous, solid oral dosage forms As such, further forms are described herein.
  • Typical oral dosage forms of the invention are prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms e.g., powders, tablets, capsules, and caplets
  • micro crystalline cellulose diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free flowing form such as powder or granules, optionally mixed with an excipient. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms of the invention include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pregelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions of the invention is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL PH 101 , AVICEL PH 103 AVICEL RC 581, AVICEL PH 105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof.
  • An specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC 581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL PH 103 and Starch 1500 LM.
  • Disintegrants are used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms of the invention.
  • the amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pregelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g. , peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • calcium stearate e.g., magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc
  • hydrogenated vegetable oil e.g. , peanut oil, cottons
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB O SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • AEROSIL 200 a syloid silica gel
  • a coagulated aerosol of synthetic silica marketed by Degussa Co. of Piano, TX
  • CAB O SIL a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA
  • lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • Active ingredients such as the compounds of the invention can be any active ingredients.
  • hydropropylmethyl cellulose other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients of the invention.
  • the invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled release.
  • controlled release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts.
  • the use of an optimally designed controlled release preparation in medical treatment is characterized by a minimum of drug substance being employed to treat or control the condition in a minimum amount of time.
  • Advantages of controlled release formulations include extended activity of the drug, reduced dosage frequency, and increased subject compliance.
  • controlled release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • the drug may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used (see, Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al, Surgery 88:507 (1980); Saudek et al, N. Engl. J. Med. 321 :574 (1989)).
  • polymeric materials can be used.
  • a controlled release system can be placed in a subject at an appropriate site determined by a practitioner of skill, i.e., thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release, vol. 2, pp. 115-138 (1984)). Other controlled release systems are discussed in the review by Langer (Science 249: 1527-1533 (1990)).
  • the active ingredient can be dispersed in a solid inner matrix, e.g.
  • parenteral dosage forms can be administered to subjects by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses subjects' natural defenses against contaminants, parenteral dosage forms are, in one embodiment, sterile or capable of being sterilized prior to administration to a subject. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
  • Suitable vehicles that can be used to provide parenteral dosage forms of the invention are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and nonaqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • Transdermal, topical, and mucosal dosage forms include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton PA (1980, 1990 & 2000); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985).
  • transdermal dosage forms include "reservoir type” or “matrix type” patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
  • Suitable excipients e.g., carriers and diluents
  • other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed by this invention are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
  • excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane- 1,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are non-toxic and pharmaceutically acceptable.
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton PA (1980, 1990 & 2000).
  • penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
  • Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide;
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery enhancing or penetration enhancing agent.
  • Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
  • the compounds of the present invention act on enzymes called cyclophilins and inhibit their catalytic activity and in another aspect, provided are methods to inhibit cyclophilins comprising administering a compound or composition provided herein to a subject in need thereof.
  • Cyclophilins occur in a wide variety of different organisms, including human, yeast, bacteria, protozoa, metazoa, insects, plants, or viruses. In the case of infectious organisms, inhibition of the cyclophilin catalytic activity by
  • compounds of the present invention often results in an inhibitory effect on the organism. Furthermore, in humans the catalytic activity of cyclophilins plays a role in many different disease situations. Inhibition of this catalytic activity is often associated to a therapeutic effect. Therefore, certain compounds of the present invention can be used for the treatment of infections including that by HCV and HIV (described further below) as well as fungal pathogens, protozoan and metazoan parasites. In addition, certain compounds of the present invention can be used to treat neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and neuropathies. Another use of the compounds of the present invention is protection against tissue damage associated to ischemia and reperfusion such as paralytic damage after spinal cord or head injuries or cardiac damage after myocardial infarct.
  • the compounds of the present invention induce regenerative processes such as that of hair, liver, gingiva, or nerve tissue damaged or lost due to injury or other underlying pathologies, such as damage of the optical nerve in glaucoma.
  • the present invention provides a method inhibiting cyclophilin in a cell comprising the administration to said cell of an effective amount of a compound of formula (I) as defined above or a pharmaceutically acceptable salt or solvate thereof.
  • Certain compounds of the present invention can be used to treat chronic inflammatory and autoimmune diseases.
  • immunosuppressants certain compounds of formula (I) are useful when administered for the prevention of immune-mediated tissue or organ graft rejection.
  • the regulation of the immune response by the compounds of the invention would also find utility in the treatment of autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosis, hyperimmunoglobulin E, Hashimoto's thyroiditis, multiple sclerosis, progressive systemic sclerosis, myasthenia gravis, type I diabetes, uveitis, allergic encephalomyelitis, glomerulonephritis.
  • Further uses include the treatment and prophylaxis of inflammatory and hyperproliferative skin diseases and cutaneous manifestations of immunologically-mediated illnesses, such as psoriasis, atopic dermatitis, contact dermatitis and further eczematous dermatitises, seborrhoeic dermatitis, Lichen planus, Pemphigus, bullous pemphigoid, Epidermolysis bullosa, urticaria, angioedemas, vasculitides, erythemas, cutaneous eosinophilias, Lupus erythematosus, acne and Alopecia areata; various eye diseases (autoimmune and otherwise) such as keratoconjunctivitis, vernal conjunctivitis, keratitis, herpetic keratitis, conical cornea, dystrophia epithelialis corneae, corneal leukoma, ocular pemphigus, Mooren's ulcer, Scleriti
  • hyperproliferative vascular diseases such as intimal smooth muscle cell hyperplasia, restenosis and vascular occlusion, particularly following biologically- or mechanically-mediated vascular injury can be treated or prevented by the compounds of the invention.
  • Other treatable conditions would include but are not limited to ischemic bowel diseases; inflammatory bowel diseases, necrotizing enterocolitis, intestinal lesions associated with thermal burns and leukotriene B4-mediated diseases; intestinal inflammations/allergies such as Coeliac diseases, proctitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease and ulcerative colitis; food-related allergic diseases which have symptomatic manifestations remote from the gastro-intestinal tract (e.g., migraine, rhinitis and eczema); renal diseases such as interstitial nephritis, Goodpasture's syndrome, hemolytic-uremic syndrome and diabetic nephropathy; nervous diseases such as multiple myositis, Guillain-Bar
  • radiculopathy endocrine diseases such as hyperthyroidism and Basedow's disease;
  • hematic diseases such as pure red cell aplasia, aplastic anemia, hypoplastic anemia, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, agranulocytosis, pernicious anemia, megaloblastic anemia and anerythroplasia; bone diseases such as osteoporosis; respiratory diseases such as sarcoidosis, fibroid lung and idiopathic interstitial pneumonia; skin disease such as dermatomyositis, leukoderma vulgaris, ichthyosis vulgaris, photoallergic sensitivity and cutaneous T cell lymphoma; circulatory diseases such as arteriosclerosis, atherosclerosis, aortitis syndrome, polyarteritis nodosa and myocardosis; collagen diseases such as scleroderma, Wegener's granuloma and Sjogren's syndrome; adiposis; eosinophilic fasciitis; periodontal disease such as lesions
  • hypobaropathy disease caused by histamine or leukotriene-C4 release
  • Behcet's disease such as intestinal-, vasculo- or neuro-Behcet's disease, and also Behcet's which affects the oral cavity, skin, eye, vulva, articulation, epididymis, lung, kidney and so on.
  • the compounds of the invention are useful for the treatment and prevention of hepatic disease such as immunogenic diseases (for example, chronic autoimmune liver diseases such as the group consisting of autoimmune hepatitis, primary biliary cirrhosis and sclerosing cholangitis), partial liver resection, acute liver necrosis, cirrhosis (such as alcoholic cirrhosis) and hepatic failure such as fulminant hepatic failure, late-onset hepatic failure and acute liver failure or chronic liver diseases.
  • immunogenic diseases for example, chronic autoimmune liver diseases such as the group consisting of autoimmune hepatitis, primary biliary cirrhosis and sclerosing cholangitis
  • partial liver resection such as the group consisting of autoimmune hepatitis, primary biliary cirrhosis and sclerosing cholangitis
  • partial liver resection such as the group consisting of autoimmune hepatitis, primary biliary cirr
  • immunocompromised patients e.g. patients with HIV or AIDS.
  • the present invention provides methods of using a compound or composition of the invention for the treatment or prevention of a viral infection in a subject in need thereof.
  • the methods generally comprise the step of administering to the subject an effective amount of the compound or composition to treat or prevent the viral infection.
  • the viral infection is HCV infection or HIV infection, or HCV and HIV co-infection.
  • the subject can be any subject infected with, or at risk for infection with, HCV. Infection or risk for infection can be determined according to any technique deemed suitable by the practitioner of skill in the art. In certain embodiments, subjects are humans infected with HCV. In one
  • a method for inhibiting the replication of HCV comprising contacting HCV-infected cells with an effective amount of a compound of formula (I) as defined above, or a pharmaceutically salt or solvate thereof.
  • the HCV can be any HCV known to those of skill in the art. There are at least six genotypes and at least 50 subtypes of HCV currently known to those of skill in the art.
  • the HCV can be of any genotype or subtype known to those of skill.
  • the HCV is of a genotype or subtype not yet characterized.
  • the subject is infected with HCV of a single genotype. In certain embodiments, the subject is infected with HCV of multiple subtypes or multiple genotypes.
  • the HCV is genotype 1 and can be of any subtype.
  • the HCV is subtype la, lb or lc. It is believed that HCV infection of genotype 1 responds poorly to current interferon therapy. Methods of the present invention can be advantageous for therapy of HCV infection with genotype 1.
  • the HCV is other than genotype 1.
  • the HCV is genotype 2 and can be of any subtype.
  • the HCV is subtype 2a, 2b or 2c.
  • the HCV is genotype 3 and can be of any subtype.
  • the HCV is subtype 3 a or 3b.
  • the HCV is genotype 4 and can be of any subtype.
  • the HCV is subtype 4a.
  • the HCV is genotype 5 and can be of any subtype.
  • the HCV is subtype 5a.
  • the HCV is genotype 6 and can be of any subtype.
  • the HCV is subtype 6a, 6b, 7b, 8b, 9a or 11a. See, e.g., Simmonds, 2004, J Gen Virol. 85:3173-88; Simmonds, 2001, J Gen. Virol, 82, 693-712, the contents of which are incorporated by reference in their entirety and relied upon.
  • the subject has never received therapy or prophylaxis for HCV infection.
  • the subject has previously received therapy or prophylaxis for HCV infection.
  • the subject has not responded to HCV therapy. Indeed, under current interferon therapy, up to 50% or more HCV subjects do not respond to therapy.
  • the subject can be a subject that received therapy but continued to suffer from viral infection or one or more symptoms thereof.
  • the subject can be a subject that received therapy but failed to achieve a sustained virologic response.
  • the subject has received therapy for HCV infection but has failed show a 2 logio decline in HCV RNA levels after 12 weeks of therapy.
  • the subject is a subject that discontinued HCV therapy because of one or more adverse events associated with the therapy.
  • the subject is a subject where current therapy is not indicated. For instance, certain therapies for HCV are associated with neuropsychiatric events.
  • Interferon (IFN)-alfa plus ribavirin is associated with a high rate of depression.
  • Depressive symptoms have been linked to a worse outcome in a number of medical disorders.
  • Life-threatening or fatal neuropsychiatric events including suicide, suicidal and homicidal ideation, depression, relapse of drug addiction/overdose, and aggressive behavior have occurred in subjects with and without a previous psychiatric disorder during HCV therapy.
  • Interferon-induced depression is a limitation for the treatment of chronic hepatitis C, especially for subjects with psychiatric disorders.
  • Psychiatric side effects are common with interferon therapy and responsible for about 10% to 20% of discontinuations of current therapy for HCV infection.
  • the present invention provides methods of treating or preventing HCV infection in subjects where the risk of neuropsychiatric events, such as depression, contraindicates treatment with current HCV therapy.
  • the present invention also provides methods of treating or preventing HCV infection in subjects where a neuropsychiatric event, such as depression, or risk of such indicates discontinuation of treatment with current HCV therapy.
  • the present invention further provides methods of treating or preventing HCV infection in subjects where a neuropsychiatric event, such as depression, or risk of such indicates dose reduction of current HCV therapy.
  • ribavirin is toxic to red blood cells and is associated with hemolysis.
  • the present invention also provides methods of treating or preventing HCV infection in subjects hypersensitive to interferon or ribavirin, or both, subjects with a hemoglobinopathy, for instance thalassemia major subjects and sickle-cell anemia subjects, and other subjects at risk from the hematologic side effects of current therapy.
  • a hemoglobinopathy for instance thalassemia major subjects and sickle-cell anemia subjects
  • the subject has received HCV therapy and
  • the subject has received therapy and continues to receive that therapy along with administration of a method of the invention.
  • the methods of the invention can be co-administered with other therapy for HCV according to the judgment of one of skill in the art.
  • the methods or compositions of the invention can be co-administered with a reduced dose of the other therapy for HCV.
  • the present invention provides methods of treating a subject that is refractory to treatment with interferon. For instance, in some embodiments, in some
  • the subject can be a subject that has failed to respond to treatment with one or more agents selected from the group consisting of interferon, interferon a, pegylated interferon a, interferon plus ribavirin, interferon a plus ribavirin and pegylated interferon a plus ribavirin.
  • the subject can be a subject that has responded poorly to treatment with one or more agents selected from the group consisting of interferon, interferon a, pegylated interferon a, interferon plus ribavirin, interferon a plus ribavirin and pegylated interferon a plus ribavirin.
  • the present invention provides methods of treating HCV infection in subjects that are pregnant or might get pregnant since current therapy is also contraindicated in pregnant women.
  • the methods or compositions of the invention are administered to a subject following liver transplant. Hepatitis C is a leading cause of liver transplantation in the U.S, and many subjects that undergo liver transplantation remain HCV positive following transplantation.
  • the present invention provides methods of treating such recurrent HCV subjects with a compound or composition of the invention.
  • the present invention provides methods of treating a subject before, during or following liver transplant to prevent recurrent HCV infection.
  • Cyclosporine compounds of general formula (I) can be particularly useful in the prophylaxis and treatment of retrovirus diseases and more particularly of AIDS and of syndromes associated with AIDS.
  • Prophylaxis is understood to mean in particular the treatment of subjects who have been exposed to HIV viruses, in particular asymptomatic seropositives who present the risk of developing the disease in the months or years to come after the primary infection.
  • the cyclosporine compounds of general formula (I) according to the invention can display an anti-retrovirus activity at concentrations devoid of any cytotoxic or cytostatic effect.
  • the subject can be any subject infected with, or at risk for infection with, HIV. Infection or risk for infection can be determined according to any technique deemed suitable by the practitioner of skill in the art. In certain embodiment, subjects are humans infected with HIV.
  • the HIV can be any HIV known to those of skill in the art.
  • the subject has never received therapy or prophylaxis for HIV infection.
  • the subject has previously received therapy or prophylaxis for HIV infection.
  • the subject has not responded to HIV therapy.
  • the subject can be a subject that received therapy but continued to suffer from viral infection or one or more symptoms thereof.
  • the subject can be a subject that received therapy but failed to achieve a sustained virologic response.
  • the subject is a subject that discontinued HIV therapy because of one or more adverse events associated with the therapy.
  • the subject is a subject where current therapy is not indicated.
  • the subject has received HIV therapy and discontinued that therapy prior to administration of a method of the invention.
  • the subject has received therapy and continues to receive that therapy along with administration of a method of the invention.
  • the methods of the invention can be co-administered with other therapy for HIV according to the judgment of one of skill in the art.
  • the methods or compositions of the invention can be co-administered with a reduced dose of the other therapy for HIV.
  • the present invention provides methods of treating a subject that is refractory to treatment for HIV.
  • the subject can be a subject that has failed to respond to treatment with one or more therapeutic agents for HIV.
  • the subject can be a subject that has responded poorly to treatment with one or more therapeutic agents for HIV.
  • the subject has, or is at risk for, co-infection of HCV with HIV.
  • 30% of HIV subjects are co-infected with HCV and evidence indicates that people infected with HIV have a much more rapid course of their hepatitis C infection.
  • the methods of the invention can be used to treat or prevent HCV infection in such subjects. It is believed that elimination of HCV in these subjects will lower mortality due to end-stage liver disease. Indeed, the risk of progressive liver disease is higher in subjects with severe AIDS-defining immunodeficiency than in those without. See, e.g., Lesens et ah, 1999, J Infect Dis 179: 1254-1258.
  • the present invention provides methods of treating or preventing HIV infection and HCV infection in subjects in need thereof.
  • doses are from about 1 to about 1500 mg per day for an adult, or from about 50 to about 1300 mg per day or from about 100 to 1 100 mg per day for an adult. In one embodiment, dose rates are from about 250 to about 1000 mg per day.
  • the present invention provides methods of treating or preventing HIV and/or HCV infection in a subject by administering, to a subject in need thereof, an effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, with a high therapeutic index against HIV and/or HCV.
  • the therapeutic index can be measured according to any method known to those of skill in the art, such as the method described in the examples below.
  • the therapeutic index is the ratio of a concentration at which the compound is toxic, to the concentration that is effective against HIV and/or HCV.
  • Toxicity can be measured by any technique known to those of skill including cytotoxicity (e.g. IC 50 or IC 90 ) and lethal dose (e.g. LD 50 or LD 90 ).
  • effective concentrations can be measured by any technique known to those of skill including effective concentration (e.g. EC 50 or EC 90 ) and effective dose (e.g. ED 50 or ED 90 ).
  • the amount of the compound or composition of the invention which will be effective in the prevention or treatment of a disorder or one or more symptoms thereof will vary with the nature and severity of the disease or condition, and the route by which the active ingredient is administered.
  • the frequency and dosage will also vary according to factors specific for each subject depending on the specific therapy (e.g., therapeutic or prophylactic agents) administered, the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the subject.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • Exemplary doses of a composition include milligram or microgram amounts of the active compound per kilogram of subject or sample weight (e.g., about 10 micrograms per kilogram to about 50 milligrams per kilogram, about 100 micrograms per kilogram to about 25 milligrams per kilogram, or about 100 microgram per kilogram to about 10 milligrams per kilogram).
  • the dosage administered to a subject is typically 0.140 mg/kg to 3 mg/kg of the subject's body weight, based on weight of the active compound.
  • the dosage administered to a subject is between 0.20 mg/kg and 2.00 mg/kg, or between 0.30 mg/kg and 1.50 mg/kg of the subject's body weight.
  • the recommended daily dose range of a composition of the invention for the conditions described herein lie within the range of from about 0.1 mg to about 1500 mg per day, given as a single once-a-day dose or as divided doses throughout a day.
  • the daily dose is administered twice daily in equally divided doses.
  • a daily dose range should be from about 50 mg to about 1300 mg per day, more specifically, between about 100 mg and about 1100 mg per day, or even more specifically between about 250 and about 1000 mg per day. It may be necessary to use dosages of the active ingredient outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art.
  • the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with subject response.
  • treatment or prevention can be initiated with one or more loading doses of a compound or composition of the invention followed by one or more maintenance doses.
  • a dose of a compound or composition of the invention can be administered to achieve a steady-state concentration of the active ingredient in blood or serum of the subject.
  • the steady-state concentration can be determined by measurement according to techniques available to those of skill or can be based on the physical characteristics of the subject such as height, weight and age.
  • the present invention provides unit dosages comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, in a form suitable for administration. Such forms are described in detail above.
  • the unit dosage comprises 1 to 1500 mg, 5 to 250 mg or 10 to 50 mg active ingredient.
  • the unit dosages comprise about 1, 5, 10, 25, 50, 100, 125, 250, 500 or 1000 mg active ingredient.
  • Such unit dosages can be prepared according to techniques familiar to those of skill in the art.
  • the present invention provides methods of treatment or prevention that comprise the administration of a second agent effective for the treatment or prevention of HIV and/or HCV infection in a subject in need thereof.
  • the second agent can be any agent known to those of skill in the art to be effective for the treatment or prevention of the HIV and/or HCV infection.
  • the second agent can be a second agent presently known to those of skill in the art, or the second agent can be second agent later developed for the treatment or prevention of HIV and/or HCV.
  • the second agent is presently approved for the treatment or prevention of HIV and/or HCV.
  • a compound of the invention is administered in combination with one second agent, for example a HCV agent.
  • a second agent is administered in combination with two second agents.
  • a second agent is administered in combination with two or more second agents.
  • a second HCV agent include interferon, pegylated interferon, ribavirin, a protease inhibitor such as telaprevir, boceprevir or ITMN-191or, or a polymerase inhibitor such as R-7128.
  • a compound of the invention is provided in combination with two other HCV agents (for example pegylated interferon and ribavirin, pegylated interferon and a protease inhibitor, pegylated interferon and a polymerase inhibitor).
  • HCV agents for example pegylated interferon and ribavirin, pegylated interferon and a protease inhibitor, pegylated interferon and a polymerase inhibitor.
  • three other HCV agents for example pegylated interferon, ribavirin and a protease inhibitor; pegylated interferon, ribavirin and a polymerase inhibitor; ribavirin, a protease inhibitor and a polymerase inhibitor.
  • kits for use in methods of treatment or prophylaxis of HIV and/or HCV infection can include a pharmaceutical compound or composition of the invention and instructions providing information to a health care provider regarding usage for treating or preventing a bacterial infection. Instructions may be provided in printed form or in the form of an electronic medium such as a floppy disc, CD, or DVD, or in the form of a website address where such instructions may be obtained.
  • a unit dose of a compound or composition of the invention can include a dosage such that when administered to a subject, a therapeutically or prophylactically effective plasma level of the compound or composition can be maintained in the subject for at least 1 day.
  • a compound or composition of the invention can be included as a sterile aqueous pharmaceutical composition or dry powder (e.g., lyophilized) composition.
  • suitable packaging refers to a solid matrix or material customarily used in a system and capable of holding within fixed limits a compound or composition of the invention suitable for administration to a subject.
  • materials include glass and plastic (e.g., polyethylene, polypropylene, and polycarbonate) bottles, vials, paper, plastic, and plastic-foil laminated envelopes and the like. If e-beam sterilization techniques are employed, the packaging should have sufficiently low density to permit sterilization of the contents.
  • Kits of the invention may also comprise, in addition to the compound or composition of the invention, second agents or compositions comprising second agents for use with compound or composition as described in the methods above.
  • Kits of the invention may also comprise, in addition to the compound or composition of the invention, second agents or compositions comprising second agents for use with compound or composition as described in the methods above.
  • the following Examples illustrate the synthesis of representative cyclosporine compounds used in the present invention and the following Reference Examples illustrate the synthesis of intermediates in their preparation. These examples are not intended, nor are they to be construed, as limiting the scope of the invention. It will be clear that the invention may be practiced otherwise than as particularly described herein. Numerous modifications and variations of the present invention are possible in view of the teachings herein and, therefore, are within the scope of the invention.
  • reaction mixture was allowed to cool to room temperature, diluted with dichloromethane and evaporated with silica gel. Purification by flash column chromatography eluting with 0-100% of a gradient mixture of 10% methanol/ethyl acetate in heptane afforded after lyophilization [4'-benzyloxy-N- methylleucine] 4 cyclosporine A (Compound A) as a white solid.
  • the HCV RNA replicon ET contains the 5' end of HCV (with the HCV Internal Ribosome Entry Site (IRES) and the first few amino acids of the HCV core protein) which drives the production of a firefly luciferase (LUC), ubiquitin, and neomycin phosphotransferase (NeoR) fusion protein. Ubiquitin cleavage releases the LUC and NeoR proteins.
  • the EMCV IRES element controls the translation of the HCV structural proteins NS3-NS5.
  • the NS3 protein cleaves the HCV polyprotein to release the mature NS3, NS4A, NS4B, NS5A and NS5B proteins that are required for HCV replication.
  • At the 3 ' end of the replicon is the authentic 3' NTR of HCV.
  • the activity of the LUC reporter is directly proportional to HCV replication levels and positive-control antiviral compounds produce a reproducible antiviral response using the LUC endpoint.
  • the compounds are dissolved in DMSO at five half-log concentrations each, ranging from either 0.03 to 3 ⁇ or 1 to 100 ⁇ .
  • Subconfluent cultures of the ET line are plated out into 96 well plates dedicated for the analysis of cell numbers (cytotoxicity) or antiviral activity and the next day the compounds are added to the appropriate wells.
  • the cells are processed 72 hours later when the cells were still subconfluent.
  • Antiviral activity is expressed as EC 50 and EC 90 , the effective concentration of compound that reduced viral replication by 50% and 90%, respectively.
  • Compound EC 50 and EC 90 values are derived from HCV RNA levels assessed as HCV RNA replicon derived LUC activity.
  • Cytotoxicity is expressed as IC 50 and IC 90 , the concentration of compound that inhibit cell viability by 50%> and 90%>, respectively.
  • Compound IC 50 and IC 90 values are calculated using a colorimetric assay as an indication of cell numbers and cytotoxicity.
  • the activity of the LUC reporter is directly proportional to HCV RNA levels in the human cell line.
  • the HCV-replicon assay is validated in parallel experiments using interferon-alpha-2b as a positive control. Cyclosporine A is tested by way of comparison. Representative compounds of the invention demonstrated activity in this assay. By way of example, compound A gave an EC 50 value of 410 nM.
  • the compounds of the present invention are also tested for antiretroviral activity against human immunodeficiency virus- 1 (HIV) using infection of the human T- lymphoblastoid cell line, CEM-SS, with the HIV strain HIV-IIIIB (Weislow et al, 1989, J Natl. Cancer Inst. 81 :577-586).
  • HIV-IIIIB Weislow et al, 1989, J Natl. Cancer Inst. 81 :577-586.
  • each experiment includes cell control wells (cells only), virus control wells (cells plus virus), drug toxicity wells (cells plus drug only), drug colorimetric control wells (drug only) as well as experimental wells (drug plus cells plus virus).
  • the assay is validated in parallel experiments using Zidovudine (3'-azido-3'-deoxythymidine or AZT) as a positive control.
  • the assay includes determinations of compound EC 50 (concentration inhibiting virus replication by 50%), IC 50 (concentration resulting in 50% inhibition of cell growth) and a selectivity index
  • D-Lys -Cs succinyl spacers bound to D-Lys -cylosporine A
  • BSA bovine serum albumin
  • a hundredfold molar excess of D-Lys -Cs dissolved in dimethyl formamide (0.6 ml) was added dropwise to the BSA under vigorous stirring.
  • the coupling reaction was performed for 2 to 3 hours at room temperature under mild stirring and the conjugate was extensively dialyzed against phosphate-buffered saline (PBS, pH 7.4). After acetone precipitation of an aliquot of the conjugated protein, no covalently bound D-Lys -Cs remained in the acetone solution and the extent of cyclosporine covalent binding was calculated.
  • Microtiter Plates were coated with D-Lys -Cs-BSA conjugate (2 pg/ml in PBS for 24 hours at 4°C). Plates were washed with Tween®/PBS and with PBS alone. To block nonspecific binding, 2% BSA/PBS (pH 7.4) was added to the wells and allowed to incubate for 2 hours at 37°C. A five-fold dilution series of the compound to be tested was made in ethanol in a separate microtiter plate. The starting concentration was 0.1 mg/mL for assays with human recombinant cyclophilin. 198 of 0.1 ⁇ g/mL cyclophilin solution was added to the microtiter immediately followed by 2 of diluted
  • cyclosporine A (used as a reference compound) or the compound of the invention.
  • the reaction between coated BSA-Cs conjugate, free cyclosporine A and cyclophilin was allowed to equilibrate overnight at 4°C.
  • Cyclophilin was detected with anti-cyclophilin rabbit antiserum diluted in 1% BSA containing PBS and incubated overnight at 4°C. Plates were washed as described above.
  • Bound rabbit antibodies were then detected by goat anti-rabbit IgG conjugated to alkaline phosphatase diluted in 1% BSA-PBS and allowed to incubate for 2 hours at 37°C. Plates were washed as described above.
  • Cells were cultured in RPMI 1640 medium, 10% fetal bovine serum, and L-Glutamine with incubation at 37°C with 5% carbon dioxide. After 1 hour of incubation the cells were stimulated with immobilized anti-CD3 (O ⁇ g/well), anti-CD28 soluble (2 ⁇ g/mL). After 6 hours the sample supernatants were harvested and stored at -80°C. 50 ⁇ ⁇ samples of supernatant were tested for IL-2 using a Luminex® 1-plex assay.
  • Mitochondrial permeability transition was determined by measuring swelling of the mitochondria induced by Ca 2+ . The procedure was adapted from the method described by Blattner et al, Analytical Biochem, Volume 295, page 220 (2001). Mitochondria were prepared from rat livers, which had been perfused with phosphate- buffered saline (PBS) to remove blood, using standard methods that utilized gentle homogenization in sucrose based buffer and then differential centrifugation to first remove cellular debris and then to pellet the mitochondria. Swelling was induced by 150 micromolar Ca 2+ (added from a concentrated solution of calcium chloride) and was monitored by measuring the scattering at 535-540 nm. Representative compounds were added 5 minutes before swelling was induced. EC50 were determined by comparing swelling with and without the compounds of the invention.
  • PBS phosphate- buffered saline

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
EP10799265A 2009-12-30 2010-12-30 Cyclosporin-analoga Withdrawn EP2519538A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US29091709P 2009-12-30 2009-12-30
PCT/US2010/062480 WO2011082289A1 (en) 2009-12-30 2010-12-30 Cyclosporine analogues

Publications (1)

Publication Number Publication Date
EP2519538A1 true EP2519538A1 (de) 2012-11-07

Family

ID=43530742

Family Applications (1)

Application Number Title Priority Date Filing Date
EP10799265A Withdrawn EP2519538A1 (de) 2009-12-30 2010-12-30 Cyclosporin-analoga

Country Status (8)

Country Link
US (1) US20120010131A1 (de)
EP (1) EP2519538A1 (de)
JP (1) JP2013516424A (de)
KR (1) KR20120125610A (de)
CN (1) CN102834409A (de)
AU (1) AU2010339460A1 (de)
CA (1) CA2785978A1 (de)
WO (1) WO2011082289A1 (de)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2011342284C1 (en) * 2010-12-15 2017-07-13 Contravir Pharmaceuticals, Inc. Cyclosporine analogue molecules modified at amino acid 1 and 3
US20140050728A1 (en) 2011-01-28 2014-02-20 Board Of Regents Of The University Of Nebraska Methods and compositions for inhibiting cyclophilin d for the treatment and prevention of obesity and kidney indications
WO2014049540A2 (en) 2012-09-29 2014-04-03 Novartis Ag Cyclic peptides and use as medicines
EP2908841A4 (de) * 2012-10-19 2016-05-25 Scynexis Inc Neue antivirale makrocyclen
EP4303288A3 (de) * 2014-07-07 2024-03-06 Nuseed Global Innovation Ltd Verfahren zur herstellung von industriellen produkten aus pflanzlichen lipiden
WO2020118715A1 (en) * 2018-12-14 2020-06-18 Waterstone Pharmaceuticals (Wuhan) Co., Ltd. Salts of scy-635 and uses thereof in medicine

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
HU196714B (en) 1984-10-04 1989-01-30 Monsanto Co Process for producing non-aqueous composition comprising somatotropin
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
IT1229203B (it) 1989-03-22 1991-07-25 Bioresearch Spa Impiego di acido 5 metiltetraidrofolico, di acido 5 formiltetraidrofolico e dei loro sali farmaceuticamente accettabili per la preparazione di composizioni farmaceutiche in forma a rilascio controllato attive nella terapia dei disturbi mentali organici e composizioni farmaceutiche relative.
PH30995A (en) 1989-07-07 1997-12-23 Novartis Inc Sustained release formulations of water soluble peptides.
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
DK0484281T4 (da) 1990-11-02 2001-02-05 Novartis Ag Cyclosporiner
US5580578A (en) 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
TW333456B (en) 1992-12-07 1998-06-11 Takeda Pharm Ind Co Ltd A pharmaceutical composition of sustained-release preparation the invention relates to a pharmaceutical composition of sustained-release preparation which comprises a physiologically active peptide.
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
IT1270594B (it) 1994-07-07 1997-05-07 Recordati Chem Pharm Composizione farmaceutica a rilascio controllato di moguisteina in sospensione liquida
CA2224381A1 (en) 1995-06-27 1997-01-16 Takeda Chemical Industries, Ltd. Method of producing sustained-release preparation
KR100441220B1 (ko) 1995-07-17 2005-02-24 체-헴 아게 항-hiv효과를갖는사이클로스포린유도체
TW448055B (en) 1995-09-04 2001-08-01 Takeda Chemical Industries Ltd Method of production of sustained-release preparation
JP2909418B2 (ja) 1995-09-18 1999-06-23 株式会社資生堂 薬物の遅延放出型マイクロスフイア
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
CA2217134A1 (en) 1996-10-09 1998-04-09 Sumitomo Pharmaceuticals Co., Ltd. Sustained release formulation
ES2221019T3 (es) 1996-10-31 2004-12-16 Takeda Chemical Industries, Ltd. Preparacion de liberacion mantenida.
WO1998027980A2 (en) 1996-12-20 1998-07-02 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
FR2757521B1 (fr) 1996-12-24 1999-01-29 Rhone Poulenc Rorer Sa Nouveaux derives de cyclosporine, leur preparation et les compositions pharmaceutiques qui les contiennent
FR2757520B1 (fr) 1996-12-24 1999-01-29 Rhone Poulenc Rorer Sa Derive de cyclosporine, sa preparation et les compositions pharmaceutiques qui le contiennent
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
FR2762843B1 (fr) 1997-04-30 1999-12-10 Rhone Poulenc Rorer Sa Nouveaux derives de cyclosporine, leur preparation et les compositions pharmaceutiques qui les contiennent
FR2772768B1 (fr) 1997-12-19 2000-01-14 Rhone Poulenc Rorer Sa Nouveau procede de preparation de derives de cyclosporine
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
KR19990085365A (ko) 1998-05-16 1999-12-06 허영섭 지속적으로 약물 조절방출이 가능한 생분해성 고분자 미립구 및그 제조방법
AU760168B2 (en) 1998-06-12 2003-05-08 Scynexis, Inc. Novel cyclosporins
FR2780061B1 (fr) 1998-06-22 2001-09-07 Rhone Poulenc Rorer Sa Nouveau procede de preparation de derives de cyclosporine
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
DE19915178A1 (de) 1999-04-03 2000-10-05 Univ Mainz Johannes Gutenberg Hepatitis C Virus Zellkultursystem
CA2583494C (en) 2004-10-01 2014-01-21 Scynexis, Inc. 3-ether and 3-thioether substituted cyclosporin derivatives for the treatment and prevention of hepatitis c infection
US7196161B2 (en) 2004-10-01 2007-03-27 Scynexis Inc. 3-ether and 3-thioether substituted cyclosporin derivatives for the treatment and prevention of hepatitis C infection
EP1931696B1 (de) 2005-09-30 2011-02-16 Scynexis, Inc. Arylalkyl- und heteroarylalkyl-derivate von cyclosporin a bei der behandlung und vorbeugung einer virusinfektion
EP1957518B1 (de) * 2005-10-26 2015-12-09 Astellas Pharma Inc. Neue zyklische peptidverbindungen
CA2748389A1 (en) * 2008-12-31 2010-07-08 Scynexis, Inc. Derivatives of cyclosporin a
EP2391376A4 (de) * 2009-01-30 2012-08-01 Enanta Pharm Inc Cyclosporin-analoga zur prävention oder behandlung von hepatitis-c-infektionen

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2011082289A1 *

Also Published As

Publication number Publication date
CN102834409A (zh) 2012-12-19
CA2785978A1 (en) 2011-07-07
US20120010131A1 (en) 2012-01-12
JP2013516424A (ja) 2013-05-13
WO2011082289A1 (en) 2011-07-07
AU2010339460A1 (en) 2012-07-19
KR20120125610A (ko) 2012-11-16

Similar Documents

Publication Publication Date Title
EP2376524B1 (de) Derivate von cyclosporin a
EP1802650B1 (de) 3-ether- und 3-thioether-substituierte ciclosporinderivate zur behandlung und prävention einer hepatitis-c-infektion
US7718767B2 (en) 3-ether and 3-thioether substituted cyclosporin derivatives for the treatment and prevention of hepatitis C infection
EP2280989B1 (de) Cyclosporinanaloge und ihre verwendung bei der behandlung von hcv-infektionen
US20110144005A1 (en) Novel cyclic peptides
US20090306033A1 (en) Novel cyclic peptides
WO2007041632A2 (en) Methods and pharmaceutical compositions for the treatment and prevention of hepatitis c infection
WO2008069917A2 (en) Novel cyclic peptides
US20120010131A1 (en) Novel cyclosporin analogues
US10738084B2 (en) Macrocycles

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120711

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20121005

R18D Application deemed to be withdrawn (corrected)

Effective date: 20130305